Pouchitis and Pouch Dysfunction Hao Wu, ,

Pouchitis and Pouch
Dys func tion
Hao Wu, MBa, Bo Shen, MDb,*
KEYWORDS
! Classification ! Complications ! Ileal pouch
! Inflammatory bowel disease
! Pouchitis ! Restorative proctocolectomy
Approximately 30% of patients with ulcerative colitis (UC) eventually require colectomy at some point in their disease course, despite advances in medical therapy.1
Restorative proctocolectomy with ileal pouch-anal anastomosis (IPAA) has become
the surgical treatment of choice for most patients with UC who fail medical therapy or
develop dysplasia, and for most patients with familial adenomatous polyposis (FAP).
The main advantage of IPAA surgery includes re-establishment of gastrointestinal
continuity and improvement of health-related quality of life. However, the trade-off
of the procedure is its high risk for the development of inflammatory and noninflammatory complications, with cumulative pouch failure rates ranging from 4% to 10%.2–6
The most common causes for pouch failure are pelvic sepsis,7,8 followed by Crohn
disease (CD) of the pouch and chronic pouchitis.9
Pouchitis is one of the most challenging disorders in IPAA. This article updates the
information on diagnosis and treatment of pouchitis.
INCIDENCE AND PREVALENCE OF POUCHITIS
Pouchitis significantly affects patients’ quality of life and long-term surgical outcome.10
Reported cumulative frequencies of pouchitis 10 to 11 years after IPAA surgery range
from 23% to 46%.11–14 It is estimated that approximately 50% of patients who have
undergone IPAA surgery for UC develop at least 1 episode of pouchitis.15 The estimated
incidence within the first 12 months after ileostomy closure was as high as 40%, as reported in a clinical trial.16 In patients with pouchitis, 70% had the initial episode during
the first 12 months after ileostomy closure.17 As the incidence of inflammatory bowel
disease (IBD), including UC, seems to be increasing, the authors expect a growing
number of patients with pouchitis or other pouch disorders in clinical practice.
This work is partially supported by a grant from BMRP, Eli and Edyth Broad Foundation.
a
Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
b
Digestive Disease Institute-Desk A31, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH
44195, USA
* Corresponding author.
E-mail address: [email protected] (B. Shen).
Gastroenterol Clin N Am 38 (2009) 651–668
doi:10.1016/j.gtc.2009.07.002
0889-8553/09/$ – see front matter ª 2009 Elsevier Inc. All rights reserved.
gastro.theclinics.com
652
Wu & Shen
ETIOLOGY AND PATHOGENESIS OF POUCHITIS
Pouchitis occurs almost exclusively in patients with underlying UC, not in patients with
FAP who undergo the same surgical procedure.18,19 It is generally believed that pouchitis results from alternations in luminal microflora (ie, dysbiosis), leading to abnormal
mucosal immune response in genetically susceptible hosts. Attempts have been
made to identify true pathogenic microbes. In a subset of patients with pouchitis,
pathogenic factors may be identified. A recent study showed that 18% of patients
seen in a specialty pouchitis clinic tested positive for Clostridium difficile toxins A or
B.20 Cytomegalovirus (CMV)21,22 and fungi (such as Candida albicans)23 have also
been implicated in pouchitis, particularly in patients with chronic antibiotic-refractory
pouchitis.
Although microbiological investigation of the bacterial communities in the gut failed
to demonstrate consistently the existence of pathogens in pouchitis, a large body of
evidence suggests that alteration in the bacteria community, ie, dysbiosis, of the
human gut likely plays a key role in the initiation and development of pouchitis. In
a culture-based study of fecal specimens in patients with UC pouches or FAP
pouches, viable sulfate-reducing bacteria were exclusively detected in pouches of
UC patients, but not in patients with FAP. Sulfate-reducing bacteria were detected
in higher numbers in active pouchitis than in those without a history of pouchitis,
past episode(s) of pouchitis, or on antibiotic therapy, and in patients with FAP.24
This particular group of bacteria was sensitive to antibiotic treatment.24 Gosselink
and colleagues25 analyzed bacteria content at the episode of pouchitis before and
during treatment with ciprofloxacin or metronidazole, and during pouchitis-free
periods, and found that, in the absence of inflammation, the pouch microbiota was
characterized by the presence of Lactobacilli and large numbers of anaerobes. During
pouchitis episodes, there was a decreased number of anaerobes, an increased
number of aerobic bacteria, lower numbers of Lactobacilli, and higher numbers of
Clostridium perfringens. In addition, hemolytic strains of Escherichia coli were
observed. Administration of metronidazole was shown to eradicate anaerobic microbiota including C perfringens, whereas treatment with ciprofloxacin inhibited the
growth of C perfringens and that of coliforms, including hemolytic strains of E coli.
Advances in molecular microbiology with 16S ribosomal RNA techniques have
provided a cornerstone of microbial taxonomy and made the assay of bacterial
composition in the gut community possible.26 In a case study, mucosa-associated
bacteria of the pouch were assayed using tissue biopsy samples at the time of colectomy, pouch construction, ileostomy closure, and postoperative routine pouch examination at 1, 3, and 12 months after ileostomy closure.26 The pouch microbiota were
similar to the normal colon microbiota except for the presence of clones with
sequences resembling those of the C perfringens group and Turicibacter. The bacterial composition differed between the 2 patients studied and the microbiota changed
with time, suggesting that the composition is not stable during the first year of ileostomy closure.26 Komanduri and colleagues27 studied pouch biopsy specimens from
5 patients with active pouchitis and 15 patients with normal pouches, using a fingerprinting technique. The study showed mucosa-associated microbiota patterns unique
to each individual. Moreover, specific bacterial amplicons were unique to active pouchitis mucosa: clostridial cluster XIVa, Enterobacteriaceae, and Streptococci were
associated with control pouches. The persistence of Fusobacter and enteric species
associated with the disease state was also shown.
Alterations in innate and adaptive mucosal immunity in the pouch and pouchitis have
been reported.28–33 An increased bacterial permeability was associated with duration of
Ileal Pouch Disorders
having a pouch with mucosal adaptive changes in an ex vivo study.28 UC patients
with backwash ileitis were shown to have impaired barrier function in the future
course of the IPAA.34 Toll-like receptors (TLR) serve an important immune and nonimmune function in human intestinal epithelial cells (IEC) by binding microbial signature
molecules and triggering innate and adaptive immune responses on stimulation.35
TLRs comprise a defense line against invading pathogens challenging the IEC layer.
TLRs can trigger the secretion of antibacterial peptides, and also link innate and
adaptive immune responses of the intestinal mucosa by attracting immune cells
from the lamina propria. An aberrant TLR expression pattern has been found in
IBD.36 An immunohistochemical study showed that TLR2 expression is up-regulated
in pouchitis and TLR4 expression is increased in the normal pouch and in pouchitis
compared with the normal ileum.37 Alterations in mRNA levels of TLR3 and TLR5
were present.38 TLR3 expression was decreased significantly, whereas TLR5 expression was increased significantly in normal pouch mucosa compared with normal ileal
mucosa.38 A combined carriership of the TLR9-1237C and CD14-260T allele seemed
to be associated with development of chronic pouchitis.39
Antimicrobial peptides produced from Paneth cells and other gut epithelial cells are
an important component of innate immunity in the intestinal tract.40–43 Paneth cells
synthesize and secrete several antimicrobial peptides, including lysozyme, secretory
phospholipase 2, and human a-defensins 5 and 6 (HD5 and HD6).42–45 The copy
number of HD5 mRNA was significantly decreased in the inflamed or noninflamed
pouch compared with the normal terminal ileum.46 Tissue mRNA copies of HD5
produced by Paneth cells and b (hBD-1, 2, 3)-defensins produced by gut epithelial cells
were increased in UC and FAP pouches immediately after surgery, compared with ileum
of controls. Initially, a- and b-defensin mRNAs were higher in UC pouches than in FAP
pouches. However, the defensin expression declined in UC and FAP pouch groups and
increased again slightly in pouchitis in patients with UC. FAP pouches without pouchitis
had strong expression of hBD-1, whereas all other defensins remained at low levels.47
As in IBD, adaptive immune mechanisms for pouchitis have been extensively
studied. For example, proliferation of immature plasma cells was increased in pouchitis.32–34,48 Proinflammatory cytokines, such as tumor necrosing factor-a, are released
mostly in the inflamed mucosa by macrophages and monocytes, leading to tissue
injury, and are considered to be involved as a secondary pathophysiologic mechanism
in pouchitis.33 The production of inflammatory mediators is increased including proinflammatory cytokines,47,49–52 cell adhesion molecules,53 platelet-activating factor,54
lipoxygenase products of arachidonic acids,55,56 vascular endothelial growth factor,56
proinflammatory neuropeptides, and other mediators.50,57–59 In general, UC pouches
expressed higher levels of inflammatory cytokines than FAP pouches.60 Abnormalities
in immunoregulatory cytokines such as IL-2, interferon-g,47,61 IL-4,47,61 and IL-1047
are also observed in pouchitis. Imbalance between proinflammatory and immunoregulatory cytokines has been described in patients with pouchitis.52 However, it is likely
that those abnormalities in mucosal adaptive immunity reflect activation of nonspecific
inflammatory cascade.50
The natural history of pouchitis may mimic that of UC, starting from an acute disease
process of bacterial etiology to chronic disease of persistent inflammation. There are
similarities in clinical presentations and immunologic abnormalities between chronic
pouchitis and UC. The presence of fecal stasis in the pouch, exposure to fecal contents,
and an increased microbial load of the pouch epithelia may result in inflammatory
changes leading to morphologic alterations in the ileal pouch mucosa mimicking
the colon epithelia in UC, namely colonic metaplasia.54,62 Colonic metaplasia, characterized by villous blunting, crypt cell hyperplasia, and colon epithelium-specific
653
654
Wu & Shen
antigens such as human tropomyosin 5, may be associated with UC-like clinical
presentations.63 Colonic metaplasia seems to be associated with dysbiosis, particularly the presence of sulfate-reducing bacteria.64 It has been reported that mucosal
butyrate oxidation in pouchitis is similar to the findings in UC.65 An alteration in mucin
glycoproteins occurs in pouchitis similar to that seen in UC.66 It is possible that the
altered glycoproteins are more susceptible to enzymatic degradation by bacteria,
making the mucus barrier less effective.67
RISK FACTORS FOR POUCHITIS
Factors associated with pouchitis have been studied extensively as part of the investigation of the etiology and pathogenesis of pouchitis. In addition, the identification of risk
factors may have a direct impact on disease prevention and prognostication. Immunogenetic studies showed that genetic polymorphisms, such as those of the IL-1 receptor
antagonist68 and NOD2/CARD15,69 may increase the risk for pouchitis. Other reported
risk factors include extensive UC,70,71 the presence of backwash ileitis,17,70 precolectomy thrombocytosis,72 the presence of concurrent primary sclerosing cholangitis
(PSC)17,73,74 or arthralgia/arthropathy,75 seropositive perinuclear antineutrophil cytoplasmic antibodies (pANCA)14,76,77 or anti-CBir1 flagellin,14 being a nonsmoker,14,71,75
and the use of nonsteroidal anti-inflammatory drugs (NSAID).71,74 Acute antibioticresponsive pouchitis and chronic antibiotic-refractory pouchitis may represent different
disease processes associated with different etiopathogenetic pathways. As such,
acute and chronic pouchitis may be associated with different risk factors.71 In a recent
study of 238 patients with different phenotypes of pouchitis, antibiotic-responsive pouchitis developed in 37 pANCA-positive patients (22%) versus 6 pANCA-negative
patients (9%), and in 12 anti-CBir1–positive patients (26%) versus 31 anti-CBir1–negative patients (16%) during a median of 47 months of follow-up.14 In one report, patients
with backwash ileitis or PSC were associated with chronic pouchitis, but not with acute
pouchitis.17 Smoking was associated with acute pouchitis,14 whereas extraintestinal
manifestations,14 preoperative thrombocytosis, a long duration of IPAA,14 and postoperative surgery-related complications78 were reported to be associated with chronic
pouchitis. Smoking seems to be protective against the development of chronic pouchitis.79 These findings suggest that chronic pouchitis and UC may share similar pathogenetic pathways, as smoking has also been shown to be protective against progression
of UC.80 Patients with chronic antibiotic-refractory pouchitis, not acute pouchitis, were
associated with concurrent autoimmune disorders.81
Concurrent PSC is associated with an increased risk for backwash ileitis in patient
with UC. Furthermore, the prevalence of PSC among patients with UC needing proctocolectomy was higher than in patients with UC in general.74 Although PSC seems to
be a risk factor for pouchitis,73,74 particularly chronic pouchitis,82,83 orthotopic liver
transplantation together with post-transplant use of immunosuppressive agents
seems not to have a detrimental impact on the disease course of pouchitis.84,85
There have been discrepancies in the literature in reported risk factors associated
with pouchitis. With regard to inconsistency in the reported risk factors, there were intrainstitutional and interinstitutional variations. These variations could largely be due to
the difference in study design, sample size, diagnostic criteria used for pouchitis,
referral pattern, and statistical methods.
DIAGNOSIS OF POUCHITIS
Diagnosis of pouchitis is not always straightforward, because there are no specific
symptoms and signs. Patients with pouchitis have a wide range of clinical
Ileal Pouch Disorders
presentations, ranging from increased stool frequency, urgency, incontinence, nighttime seepage, to abdominal perianal discomfort. These symptoms, however, can be
present in other inflammatory and noninflammatory disorders of the pouch, such as
cuffitis, CD of the pouch, and irritable pouch syndrome. Therefore, the diagnosis of
pouchitis should not be solely dependent on symptom assessment. In addition,
severity of symptoms does not necessarily correlate with the degree of endoscopic
or histologic inflammation of the pouch.86,87 To complicate the matter even more,
the diagnosis of pouch disorders can resemble hitting a moving target, as the disease
process may not be static. For example, a patient may have typical pouchitis at 1
point, and may present CD of the pouch several months later. Therefore, a combined
assessment of symptoms, endoscopic, and histologic features is advocated for the
diagnosis and differential diagnosis of pouchitis.86,88 Pouch endoscopy provides the
most valuable information on the severity and extent of mucosal inflammation, backwash ileitis, CD of the pouch or cuffitis, and the presence of other abnormalities such
as polyps, strictures, sinuses, and fistula openings (Fig. 1). Although histology has
a limited role in grading the degree of pouch inflammation, it can provide valuable
information on some special features, such as granulomas, viral inclusion bodies
(for CMV infection), pyloric gland metaplasia (a sign of chronic mucosal inflammation),
and dysplasia. A diagnostic and treatment algorithm is proposed (Fig. 2).
Laboratory testing is often necessary as a part of the evaluation of patients with
pouch disorders, particular patients with chronic pouchitis. In patients with persistent
symptoms of pouchitis, celiac serology, salicylate screening, and microbiological
assays for C difficile and CMV may be performed.89 As most patients undergo
repeated or chronic antibiotic exposure, C difficile infection has been a growing
problem.20 Fecal assays of lactoferrin and calproprotectin have been evaluated for
the diagnosis and differential diagnosis of pouchitis. Quantitative90 and qualitative91
assays of lactoferrin have been used to distinguish pouchitis from normal pouches
or irritable pouch syndrome. Fecal lactoferrin may be used as an inexpensive
screening test for pouchitis.91,92 Fecal calprotectin assay had a sensitivity of 90%
and a specificity of 76.5% for the diagnosis of pouchitis in a recent study.93 However,
the diagnostic accuracy of these studies was assessed based on the comparison of
patients with pouchitis and patients with healthy pouches, whereas other inflammatory conditions such as cuffitis and CD of the pouch were not included in the prior
studies. Other laboratory tests, such as assays of fecal dimeric M2-pyruvate kinase94
and tissue proinflammatory cytokine gene scripts,95 may also be useful in distinguishing pouchitis from noninflammatory conditions of the pouch. However, laboratory
Fig.1. Endoscopy of inflammatory disorders of the pouch. (A) Pouchitis, inflammation of the
pouch body; (B) cuffitis, inflammation of the cuff; (C) Crohn disease of the pouch, ulcers at
the neoterminal ileum.
655
656
Wu & Shen
Pouchitis Confirmed By Pouch Endoscopy
Ciprofloxacin or Metronidazole x 2 wks
No Response
Response
Infrequent
Relapse
Antx-responsive
Pouchitis
Antibiotics prn
Frequent
Relapse
Metronidazole or cipro x 2 wks
Response
Antx-dependent
Pouchitis
No Response
Antx-refractory
Pouchitis
- Probiotics
- Low-dose antibiotics
- Carbon microsphere?
Cipro+Metronidazole or
Rifaximin or
Tinidazole x4 wks
Oral or Topical 5ASAs/steroids/
Immunomodulators//biol
ogic therapy?
No Response
Evaluation for secondary causes
Fig. 2. Diagnostic and treatment algorithm of pouchitis.
tests should not replace pouch endoscopy as the first-line evaluation for the diagnosis
and differential diagnosis of pouchitis.
DIFFERENTIAL DIAGNOSIS OF POUCHITIS
There are overlaps in clinical presentations between a variety of inflammatory and
noninflammatory disorders of ileal pouches (see Fig. 1). Cuffitis is considered a variant
form of UC in the rectal cuff, particularly in patients with IPAA without mucosectomy.
Patients with cuffitis often present with bloody bowel movements, which seldom occur
in conventional pouchitis. When IPAA is constructed, there are 2 techniques to be
used for the pouch-anal anastomosis, hand-sewn versus staple techniques: the
hand-sewn IPAA with mucosectomy of the anal transition zone (ATZ) mucosa (or rectal
cuff mucosa) or a stapled IPAA at the level of the anorectal ring without mucosectomy
of the ATZ. To remove the rectal mucosa as completely as possible, a mucosectomy
with hand-sewn anastomosis is necessary. This technique normally takes longer and
has a high risk for postoperative functional problems related to seepage and incontinence due to anal canal manipulation. In contrast, the stapled anastomosis is easy to
perform and is less likely to result in functional and septic complications. The preservation of ATZ is meant to optimize anal canal sensation, eliminate sphincter stretching,
and preserve normal postoperative resting and squeeze pressures.5,7,8 However, to
allow transanal insertion of the stapler head, it is usually necessary to leave a 1- to
2-cm strip of rectal cuff/ATZ mucosa that is at risk for developing symptomatic inflammation (cuffitis) or even dysplasia.
Another common inflammatory disorder of the pouch is CD. It has been speculated
that IPAA surgery with change of bowel anatomy, anastomoses, and fecal stasis
creates a ‘‘CD-friendly’’ environment. CD of the pouch can occur after IPAA intentionally performed in a selected group of patients with Crohn colitis with no small intestinal
or perianal diseases96; CD is also inadvertently found in proctocolectomy specimens
of patients with a preoperative diagnosis of UC or indeterminate colitis. De novo CD of
the pouch, by far the most common form of CD, may develop weeks to years after
Ileal Pouch Disorders
IPAA for UC or indeterminate colitis. Clinical phenotypes of CD of the pouch can be
inflammatory, fibrostenotic, or fistulizing. There are symptoms and signs that would
suggest a diagnosis of CD, particularly fibrostenotic and fistulizing CD. It is critical
to differentiate NSAID-induced ileitis/pouchitis from CD ileitis, backwash ileitis from
diffuse pouchitis. Making a diagnosis of CD of the pouch often needs a combined
assessment of symptoms, endoscopy, histology, radiography, and sometimes examination under anesthesia.
Irritable pouch syndrome is a functional disorder in patients with IPAA.97 The
disease entity has a significant negative impact on health-related quality of life. There
are great overlaps in clinical presentation between irritable pouch syndrome and pouchitis. Contributing factors for the pathophysiology of irritable pouch syndrome
include visceral hypersensitivity, enterochromaffin cell hyperplasia,98 and proximal
small bowel bacterial overgrowth. Currently, irritable pouch syndrome is a diagnosis
of exclusion. Pouch endoscopy is the diagnostic modality of choice for to distinguish
between pouchitis and irritable pouch syndrome.
Patients with surgical complications (such as pouch sinus and pouch ischemia) can
present symptoms resembling those of pouchitis. Again, pouch endoscopy is considered the first-line diagnostic modality.
CLASSIFICATION OF POUCHITIS
The natural history of pouchitis is poorly defined. Patients with initial episodes of pouchitis almost uniformly respond to antibiotic therapy. However, relapse of pouchitis is
common. Among patients with acute pouchitis, 39% had a single acute episode that
responded to antibiotic therapy; the remaining 61% developed at least 1 recurrence.99
Approximately 5% to 19% of patients with acute pouchitis develop refractory or
rapidly relapsing forms of the disease.100–102 Pouchitis likely represents a disease
spectrum from an acute, antibiotic-responsive type to a chronic, antibiotic-refractory
entity. Based on the cause, disease duration, and activity, and response to medical
therapy, pouchitis can be categorized into: (1) idiopathic versus secondary, with
causes such as NSAID use and C difficile or CMV infection; (3) acute versus chronic,
with a cut-off time of 4 weeks of persistent symptoms; (4) infrequent episodes versus
relapsing versus continuous; and (5) responsive versus refractory to antibiotic
therapy.103
Classification based on the response to antibiotic therapy is useful in clinical practice.104 Analogous to the classification of UC according to the response to or dependency on corticosteroids, pouchitis can be classified into antibiotic-responsive,
antibiotic-dependent, and antibiotic-refractory pouchitis (Table 1).75 Pouchitis can
be diffuse or patchy. Based on the distribution of inflammation, pouchitis can be categorized into diffuse pouchitis, pouchitis with backwash ileitis, pouchitis with concurrent cuffitis, and segmental pouchitis. It is now clear that pouchitis can be seen as
a heterogeneous group, with different clinical phenotypes, that may be associated
with different risk factors, pathogenetic pathways, natural courses, and outcomes.
PROPHYLAXIS OF POUCHITIS
Some of the risk factors for pouchitis are modifiable, such as avoidance of the use of
broad-spectrum antibiotics or NSAIDs. It has been speculated that small bowel bacterial overload may be common, as there is no valve mechanism between the pouch and
distal small bowel, and pouch contents can easily reflux back to the segment of small
bowel. A diet with a low quantify of poorly absorbed short-chain carbohydrates may
reduce stool frequency in patients with or without pouchitis.105
657
658
Wu & Shen
Table 1
Classification of pouchitis
Classification Based
on Disease Course
Classification Based
on Response to
AntibioticTherapy
Acute pouchitis
Antibiotic-responsive
pouchitis
Acute relapsing pouchitis
Chronic pouchitis
Antibiotic-dependent
pouchitis
Antibiotic-refractory
pouchitis
Classification
Based on Etiology
Idiopathic pouchitis (with
unidentified pathogens
or triggering factors)
Secondary pouchitis
- Clostridium difficile–
associated pouchitis
- Cytomegalovirus-associated pouchitis
- NSAID-induced
pouchitis
- Ischemic pouchitis
- Autoimmune pouchitis
Given that approximately 40% of patients develop pouchitis within the first 12
months after ileostomy closure,16 and that most patients who develop acute pouchitis
do so within the first year after IPAA,17,106 it is reasonable to consider primary or
secondary prophylaxis therapy, particularly in patients at risk.107 Efficacy and safety
of several probiotic agents have been evaluated. In a randomized trial of VSL#3 (containing viable lyophilized bacteria of 4 strains of Lactobacillus, 3 Bifidobacterium
species, and Streptococcus salivarius subsp thermophillus) for the primary prophylaxis of the initial episode of pouchitis, 2 of 20 patients (10%) in the study group
and 8 of 20 patients (40%) in the placebo group developed pouchitis within 12 months
after IPAA.16 Lactobacillus rhamnosus GG has also shown some efficacy in delaying
the first episodes of pouchitis.9,108 Although the efficacy of probiotic or prebiotic
agents in the primary prophylaxis remains to be further investigated, patients with
risk factors for pouchitis may try these agents.
In addition to primary prophylaxis, probiotic or antibiotic agents have been used as
a maintenance therapy for secondary prophylaxis in patients with relapsing pouchitis
or antibiotic-dependent pouchitis (see later discussion).
MANAGEMENT OF POUCHITIS
It is speculated that in most patients with pouchitis, the disease process is triggered
and maintained by bacterial causes from dysbiosis of commensal bacteria. However,
there are types of pouchitis that have identifiable pathogenic factors, such as C difficile,20 Candida albicans, and CMV infection, NSAID use, and pouch ischemia. Pathogen-targeted therapy is often effective.
There might be a new disease category named ‘‘autoimmune pouchopathy.’’81 The
patients often present with symptoms similar to ‘‘bacteria-associated’’ pouchitis, such
as increased stool frequency, cramps, and urgency. On endoscopy examination,
mucosal inflammation can be present in the pouch as well as a long segment of
afferent limb. Although there are currently no established diagnostic criteria, the diagnosis of autoimmune pouchopathy may be suspected if a patient has antibiotic-refractory pouchitis, concurrent autoimmune disorders (such as rheumatoid arthritis and
Ileal Pouch Disorders
Hashimoto thyroiditis), and serum autoantibodies. For patients with autoimmune
pouchopathy, treatment with oral budesonide or a low dose of immunomodulator
may be attempted.
The therapeutic effect of probiotics parallels the restoration of mucosal immune
response to altered microflora in the pouch.2,9,108–110 For example, probiotic-treated
patients were shown to have a significant increase in the percentage of regulatory
T cells.111 Changes in mucosa-associated bacteria in pouchitis were also reflected
by therapeutic administration of probiotic agents. Kuhbacher and colleagues23 conducted a double-blind, placebo-controlled trial to study the impact of a probiotic agent
containing viable lyophilized bacteria, comprising Lactobacilli, Bifidobacteria, and
Streptococci, on the dominant mucosa-associated bacteria from patients with chronic
pouchitis in remission induced by antibiotics. The mucosal microbiota was mainly detected within the epithelium and nearly all bacteria were affiliated with the Enterobacteriaceae group. Compared with the placebo group, an increase in Enterobacteriaceae
within the mucosa during the probiotic therapy was observed. Investigation of the
molecular species was performed with the construction of taxonomic group–specific
clone libraries. Using Proteobacteria/Enterobacteriaceae group–specific primers,
slight differences in terms of phylotypic composition were observed between the
placebo and probiotic groups. Enterobacter species and E coli were mainly identified.
Lactobacillus and Bifidobacterium clone libraries generated from the probiotic group
displayed a diverse spectrum of species in comparison with the 2 other experimental
groups (pretreatment remission group and placebo group). Analysis of the mucosaassociated microbiota using an electrophoretic fingerprinting technique showed that
the probiotic therapy increased the bacterial diversity in comparison with the patients
in the pretreatment remission and placebo administration groups.
Probiotics have been used as a maintenance therapy for patients with antibioticdependent pouchitis or relapsing pouchitis. A randomized trial of VSL#3 at a dose
of 6 g/d was conducted for the maintenance and secondary prophylaxis of relapse
of pouchitis, after remission was induced by oral ciprofloxacin (1000 mg/d) plus rifaximin (2000 mg/d). During the 9-month trial of 40 patients with relapsing pouchitis, 15%
in the probiotic group relapsed versus 100% in the placebo group.109 A separate
randomized trial of VSL#3 in patients with antibiotic-dependent pouchitis showed
that 17 of 20 patients (85%) in the VSL#3 group maintained clinical remission,
compared with remission in 1 of 16 patients (6%) in the placebo group.110 A metaanalysis of 5 randomized, placebo-controlled clinical trials was performed. Pooling
of the results from these trials yielded an odds ratio of 0.04 in the treatment group
in comparison with the placebo group. The benefit of probiotics in the management
of pouchitis after IPAA operation was confirmed by the meta-analysis.107 In addition,
high-dose probiotics have been used for treating pouchitis. In a study of the probiotic
agent, VSL#3, 3600 billion bacteria/d in treating mild pouchitis, 16 of 23 patients (69%)
were in remission after treatment.112
Routine use of probiotics for induction and maintenance therapy has generated
some controversy. However, some postmarket open-labeled studies reported
a much lower response rate. These outstanding results have been challenged by 2
recent postmarket open-labeled trials. In a study of 31 patients with antibiotic-dependent pouchitis treated with VSL#3 for maintenance therapy after 2 weeks of treatment
with ciprofloxacin, 25 patients (81%) had stopped the agent at 8 months, mainly
because of the lack of efficacy or development of adverse effects.113 Similar results
were reported in a separate open-labeled trial.114
Management strategies vary based on different types of pouchitis (see Fig. 2).9
Because most pouchitis is caused by bacteria, antibiotic therapy is the mainstream
659
660
Wu & Shen
therapy. For antibiotic-responsive pouchitis, the first-line therapy includes a 14-day
course of metronidazole (15–20 mg/kg/d) or ciprofloxacin (1000 mg/d).115,116 A
randomized trial of ciprofloxacin and metronidazole showed that patients treated
with ciprofloxacin experienced significantly greater reductions in disease activity
scores and fewer adverse effects than those treated with metronidazole.116 A small
randomized trial of oral rifaximin 1200 mg/d versus placebo showed only a marginal
therapeutic benefit for active pouchitis.117 Diffuse pouchitis can be associated with
backwash ileitis, particularly in patients with concurrent PSC. Combination therapy
with ciprofloxacin and metronidazole for 28 days was shown to be effective in treating
backwash ileitis in a recent open-labeled trial.118 Other agents were reported in openlabeled trials, including tetracycline, clarithromycin, amoxicillin/clavulanic acid, doxycycline, budesonide enemas, alicaforsen enemas (an antisense inhibitor of intercellular
adhesion molecule-1), leukocytapheresis,119 AST-120 (a highly adsorptive, porous,
carbon microsphere),120 and dietary supplement of short-chain carbohydrates.
The gut-specific nonabsorbable antibiotic, rifaximin, may be a candidate for maintenance therapy in patients who need long-term antibiotics. In our recent study, 51
patients began maintenance therapy with rifaximin (median dose 200 mg/d); 33
(65%) maintained remission for 3 months. Of these 33 patients, 26 (79%) successfully
continued maintenance for 6 months after beginning maintenance, 19 (58%) successfully continued for 12 months, and 2 (6%) successfully continued for 24 months.121
Treatment of chronic antibiotic-refractory pouchitis is often challenging. In fact, this
phenotype of pouchitis is one of the most common causes for pouch failure. It is
important to investigate contributing causes related to failure of antibiotic therapy.
Secondary causes of refractory disease include use of NSAIDs, concurrent C difficile,20 CMV,21,22 or fungal23 infection, celiac disease and other autoimmune disorders,
cuffitis, and CD of the pouch. Chronic pouchitis can be associated with single or
multiple small or large inflammatory polyps. Large (>1 cm) pouch polyps can occasionally cause bleeding and can be dysplastic. Endoscopic polypectomy is feasible,
which may be helpful in controlling patients’ symptoms, in conjunction with medical
therapy.122 For patients without obvious causes, treatment options include a prolonged course of combined antibiotic therapy, mesalamine, corticosteroids, immunosuppressive agents, or even biologic therapy. In open-labeled trials, combined
agents of ciprofloxacin (1000 mg/d) with rifaximin (2000 mg/d),123,124 metronidazole
(1000 mg/d),125 or tinidazole (1000–1500 mg/d) for 4 weeks were reported to be effective.126 However, maintenance of remission in this group of patients after the induction
therapy with the dual antibiotic therapy remains challenging.127
Anti-inflammatory agents, immunomodulators, and biologic therapy have been
used to treat pouchitis. These agents include bismuth carbomer enemas, short-chain
fatty acid enemas, glutamine enemas, mesalamine enemas, 6-mercaptopurine, and
infliximab. In an open-labeled study of 20 patients with chronic refractory pouchitis,
oral budesonide 9 mg/d for 8 weeks induced remission in 15 (75%).128 Biologic agents
have been used in chronic pouchitis. Infliximab was studied for treating 10 patients
with chronic refractory pouchitis complicated by ileitis, using wireless capsule endoscopy. Clinical remission was achieved in 9 patients and endoscopic (with video
capsule endoscopy and pouch endoscopy) remission.129
In summary, management of pouchitis and other pouch disorders can be difficult.
One of the contributing factors for the complexity is the surgical component, which
may not be familiar to practicing gastroenterologists. A multidisciplinary approach
involving gastroenterologists and colorectal surgeons, together with a team of gastrointestinal pathologists and gastrointestinal radiologists, has been advocated. Few
institutions have even established subspecialty pouchitis clinics.10,130 The practice
Ileal Pouch Disorders
model has been feasible and effective in treating complicated pouch disorders in
a tertiary-care setting.
SUMMARY
Pouchitis is the most common long-term complication of IPAA, which represents
a spectrum of disease processes with different clinical phenotypes, risk factors, pathogenetic pathways, natural history, and prognosis. Pouch endoscopy is the most valuable tool for diagnosis and differential diagnosis. Although most patients with
pouchitis respond favorably to antibiotic therapy, antibiotic dependency and refractory disease have posed therapeutic challenges. Secondary causes of pouchitis,
such as C difficile infection, should be evaluated.
REFERENCES
1. Dhillon S, Loftus EV Jr, Tremaine WJ, et al. The natural history of surgery for ulcerative colitis in a population-based cohort from Olmsted County, Minnesota
[abstract]. Am J Gastroenterol 2005;100:A819.
2. Sandborn WJ. Pouchitis following ileal pouch-anal anastomosis: definition, pathogenesis, and treatment. Gastroenterology 1994;107:1856–60.
3. Winther KV, Jess T, Langholz E, et al. Survival and cause-specific mortality in
ulcerative colitis - follow-up of a population-based cohort in Copenhagen
County. Gastroenterology 2003;125:1576–82.
4. Tulchinsky H, Hawley PR, Nicholls J. Long-term failure after restorative proctocolectomy for ulcerative colitis. Ann Surg 2003;238:229–34.
5. Belliveau P, Trudel J, Vasilevsky CA, et al. Ileoanal anastomosis with reservoirs:
complications and long-term results. Can J Surg 1999;42:345–52.
6. Hueting WE, Buskens E, van der Tweel I, et al. Results and complications after
ileal pouch anal anastomosis: a meta-analysis of 43 observational studies
comprising 9,317 patients. Dig Surg 2005;22:69–79.
7. Sagap I, Remzi FH, Hammel JP, et al. Factors associated with failure in
managing pelvic sepsis after ileal pouch-anal anastomosis (IPAA) – a multivariate
analysis. Surgery 2006;140:691–703 [discussion: 703–4].
8. Prudhomme M, Dehni N, Dozois RR, et al. Causes and outcomes of pouch excision after restorative proctocolectomy. Br J Surg 2006;93:82–6.
9. Nicholls RJ. Review article: ulcerative colitis-surgical indications and treatment.
Aliment Pharmacol Ther 2002;16:25–8.
10. Shen B, Fazio VW, Lashner BA, et al. Comprehensive evaluation of inflammatory
and non-inflammatory sequelae of ileal pouch-anal anastomosis. Am J Gastroenterol 2005;100:93–101.
11. Penna C, Dozois R, Tremaine W, et al. Pouchitis after ileal pouch-anal anastomosis for ulcerative colitis occurs with increased frequency in patients with associated primary sclerosing cholangitis. Gut 1996;38:234–9.
12. Fazio VW, Ziv Y, Church JM, et al. Ileal pouch-anal anastomosis complications
and function in 1005 patients. Ann Surg 1995;222:120–7.
13. Ferrante M, Declerck S, De Hertogh G, et al. Outcome after proctocolectomy
with ileal pouch-anal anastomosis for ulcerative colitis. Inflamm Bowel Dis
2008;14:20–8.
14. Fleshner P, Ippoliti A, Dubinsky M, et al. Both preoperative perinuclear antineutrophil cytoplasmic antibody and anti-CBir1 expression in ulcerative colitis
patients influence pouchitis development after ileal pouch-anal anastomosis.
Clin Gastroenterol Hepatol 2008;6:561–8.
661
662
Wu & Shen
15. Stocchi L, Pemberton JH. Pouch and pouchitis. Gastroenterol Clin North Am
2001;30:223–41.
16. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind placebo controlled trial. Gastroenterology 2003;
124:1202–9.
17. Abdelrazeq AS, Kandiyil N, Botterill ID, et al. Predictors for acute and chronic
pouchitis following restorative proctocolectomy for ulcerative colitis. Colorectal
Dis 2008;10:805–13.
18. Penna C, Tiret E, Kartheuser A, et al. Function of ileal J pouch-anal anastomosis in patients with familial adenomatous polyposis. Br J Surg 1993;80:
765–7.
19. Tjandra JJ, Fazio VW, Church JM, et al. Similar functional results after restorative
proctocolectomy in patients with familial adenomatous polyposis and mucosal
ulcerative colitis. Am J Surg 1993;165:322–5.
20. Shen B, Jiang Z-D, Fazio VW, et al. Clostridium difficile infection in patients with
ileal pouch-anal anastomosis. Clin Gastroenterol Hepatol 2008;6:782–8.
21. Munoz-Juarez M, Pemberton JH, Sandborn WJ, et al. Misdiagnosis of specific
cytomegalovirus infection of ileoanal pouch as a refractory idiopathic chronic
pouchitis. Report of two cases. Dis Colon Rectum 1999;42:117–20.
22. Mooka D, Furth EE, MacDermott RP, et al. Pouchitis associated with primary
cytomegalovirus infection. Am J Gastroenterol 1998;93:264–6.
23. Ku¨hbacher T, Ott SJ, Helwig U, et al. Bacterial and fungal microbiota in relation
to probiotic therapy (VSL#3) in pouchitis. Gut 2006;55:833–41.
24. Ohge H, Furne JK, Springfield J, et al. Association between fecal hydrogen
sulfide production and pouchitis. Dis Colon Rectum 2005;48:469–75.
25. Gosselink MP, Schouten WR, van Lieshout LM, et al. Eradication of pathogenic
bacteria and restoration of normal pouch flora: comparison of metronidazole and
ciprofloxacin in the treatment of pouchitis. Dis Colon Rectum 2004;47:1519–25.
26. Falk A, Olsson C, Ahrne S, et al. Ileal pelvic pouch microbiota from two former
ulcerative colitis patients, analysed by DNA-based methods, were unstable
over time and showed the presence of Clostridium perfringens. Scand J Gastroenterol 2007;42:973–85.
27. Komanduri S, Gillevet PM, Sikaroodi M, et al. Dysbiosis in pouchitis: evidence of
unique microfloral patterns in pouch inflammation. Clin Gastroenterol Hepatol
2007;5:352–60.
28. Kroesen AJ, Leistenschneider P, Lehmann K, et al. Increased bacterial permeation in long-lasting ileoanal pouches. Inflamm Bowel Dis 2006;12:736–44.
29. DeSilva HJ, Jones M, Prince C, et al. Lymphocyte and macrophage subpopulations in pelvic ileal reservoirs. Gut 1991;32:1160–5.
30. Hirata I, Berrebi G, Austin LL, et al. Immunohistological characterization of intraepithelial and lamina propria lymphocytes in control ileum and colon and inflammatory bowel disease. Dig Dis Sci 1986;31:593–603.
31. Stallmach A, Schafer F, Hoffman S, et al. Increased state of activation of CD4
positive T cells and elevated interferon gamma production in pouchitis. Gut
1998;43:499–505.
32. Thomas PD, Forbes A, Nicholls RJ, et al. Altered expression of the lymphocyte
activation markers CD30 and CD27 in patients with pouchitis. Scand J Gastroenterol 2001;36:258–64.
33. Goldberg PA, Herbst F, Beckett CG, et al. Leukocyte typing, cytokine expression
and epithelial turnover in the ileal pouch in patients with ulcerative colitis and
familial adenomatous polyposis. Gut 1996;38:549–53.
Ileal Pouch Disorders
34. Kroesen AJ, Dullat S, Schulzke JD, et al. Permanently increased mucosal
permeability in patients with backwash ileitis after ileoanal pouch for ulcerative
colitis. Scand J Gastroenterol 2008;43:704–11.
35. Akira S, Takeda A. Toll like receptors signalling. Nat Rev Immunol 2004;4(7):
499–511.
36. Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression
of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect
Immun 2000;68(12):7010–7.
37. Toiyama T, Araki T, Yoshiyama S, et al. The expression patterns of toll-like receptors in the ileal pouch mucosa of postoperative ulcerative colitis patients. Surg
Today 2006;36:287–90.
38. Heuschen G, Leowardi C, Hinz U, et al. Differential expression of toll-like
receptor 3 and 5 in ileal pouch mucosa of ulcerative colitis patients. Int J Colorectal Dis 2007;22:293–301.
39. Lammers KM, Ouburg S, Morre SA, et al. Combined carriership of TLR9 -1237C
and CD14 -260T alleles enhances the risk for developing chronic relapsing pouchitis. World J Gastroenterol 2005;11:7323–9.
40. Ayabe T, Satchell DP, Wilson CL, et al. Secretion of microbicidal alpha-defensins
by intestinal Paneth cells in response to bacteria. Nat Immunol 2000;1:113–8.
41. Salzman NH, Ghosh D, Huttner KM, et al. Protection against enteric salmonellosis in
transgenic mice expressing a human intestinal defensin. Nature 2003;422:522–6.
42. Ouellette A, Bevins CL. Paneth cell defensins and innate immunity of the small
bowel. Inflamm Bowel Dis 2001;7:43–50.
43. Porter EM, Bevins CL, Ghosh D, et al. The multifaceted Paneth cell. Cell Mol Life
Sci 2002;59:156–70.
44. Ghosh D, Porter EM, Shen B, et al. Paneth cell trypsin is the processing enzyme
for human defensin-5. Nat Immunol 2002;3:583–90.
45. Porter E, van Dam E, Valore E, et al. Broad spectrum antimicrobial activity of
human intestinal defensin 5. Infect Immun 1997;65:2396–401.
46. Wehkamp J, Salzman NH, Porter E, et al. Decreased Paneth cell defensins and
antimicrobial activity in ileal Crohn’s disease. Proc Natl Acad Sci U S A 2005;
102:18129–34.
47. Kiehne K, Brunke G, Wegner F, et al. Defensin expression in chronic pouchitis in
patients with ulcerative colitis or familial adenomatous polyposis coli. World J
Gastroenterol 2006;12:1056–62.
48. Hirata N, Oshitani N, Kamata N, et al. Proliferation of immature plasma cells in
pouchitis mucosa in patients with ulcerative colitis. Inflamm Bowel Dis 2008;
14:1084–90.
49. Patel RT, Bain I, Youngs D, et al. Cytokine production in pouchitis is similar to that
in ulcerative colitis. Dis Colon Rectum 1995;38:831–7.
50. Schmidt C, Giese T, Ludwig B, et al. Increased cytokine transcripts in pouchitis
reflect the degree of inflammation but not the underlying entity. Int J Colorectal
Dis 2006;21:419–26.
51. Gionchetti P, Campieri M, Belluzzi A, et al. Mucosal concentrations of interleukin1b, interleukin-6, interleukin-8, and tumor necrosis factor-a in pelvic ileal
pouches. Dig Dis Sci 1994;39:1525–31.
52. Bulois P, Tremaine WJ, Maunoury V, et al. Pouchitis is associated with mucosal
imbalance between interleukin-8 and interleukin-10. Inflamm Bowel Dis 2000;6:
157–64.
53. Patel RT, Pall AA, Adu D, et al. Circulating soluble adhesion molecules in inflammatory bowel disease. Eur J Gastroenterol Hepatol 1995;7:1037–41.
663
664
Wu & Shen
54. Chaussade S, Denizot Y, Valleur P, et al. Presence of PAF-acether in stool of
patients with pouch ileoanal anastomosis and pouchitis. Gastroenterology
1991;100:1509–14.
55. Gertner DJ, Rampton DS, Madden MV, et al. Increased leukotriene B4 release
from ileal pouch mucosa in ulcerative colitis compared with familial adenomatous polyposis. Gut 1994;35:1429–32.
56. Romano M, Cuomo A, Tuccillo C, et al. Vascular endothelial growth factor and
cyclooxygenase-2 are overexpressed in ileal pouch-anal anastomosis. Dis
Colon Rectum 2007;50:650–9.
57. Stucchi AF, Shebani KO, Leeman SE, et al. A neurokinin 1 receptor antagonist
reduces an ongoing ileal pouch inflammation and the response to a subsequent
inflammatory stimulus. Am J Physiol Gastrointest Liver Physiol 2003;285:
G1259–67.
58. Stallmach A, Chan CC, Ecker K-W, et al. Comparable expression of matrix
metalloproteinases 1 and 2 in pouchitis and ulcerative colitis. Gut 2000;47:
415–22.
59. Ulisse S, Gionchetti P, Alo S, et al. Expression of cytokine, inducible nitric oxide
synthase, matrix metalloproteinases in pouchitis: effects of probiotic therapy. Am
J Gastroenterol 2001;96:2691–6.
60. Leal RF, Coy CS, Ayrizono ML, et al. Differential expression of pro-inflammatory
cytokines and a pro-apoptotic protein in pelvic ileal pouches for ulcerative colitis
and familial adenomatous polyposis. Tech Coloproctol 2008;12:33–8.
61. Thomas PD, Forbes A, Nicholls RJ, et al. Increased mucosal IFN-g production in
pouchitis despite normal functional responses of isolated CD4 cells [abstract].
Gut 1999;44(Suppl 4):32A.
62. Shepherd NA, Healey CJ, Warren BF, et al. Distribution of mucosal morphology
and an assessment of colonic phenotype change in the pelvic ileal reservoir. Gut
1993;34:101–5.
63. Biancone L, Palmieri G, Lombardi A, et al. Tropomyosin expression in the ileal
pouch: a relationship with the development of pouchitis in ulcerative colitis.
Am J Gastroenterol 2003;98:2719–26.
64. Coffey JC, Rowan F, Burke J, et al. Pathogenesis of and unifying hypothesis
for idiopathic pouchitis. Am J Gastroenterol 2009;104:1013–23.
65. De Preter V, Bulteel V, Suenaert P, et al. Pouchitis, similar to active ulcerative
colitis, is associated with impaired butyrate oxidation by intestinal mucosa.
Inflamm Bowel Dis 2009;15:335–40.
66. Tysk C, Riedesel H, Lindberg E, et al. Colonic glycoproteins in monozygote twins
with inflammatory bowel disease. Gastroenterology 1991;100:1509–14.
67. Merrett MN, Soper N, Mortensen N, et al. Intestinal permeability in the ileal
pouch. Gut 1996;39:226–30.
68. Carter K, Di Giovine FS, Cox A, et al. The interleukin 1 receptor antagonist gene
allele 2 as a predictor of pouchitis following colectomy and IPAA in ulcerative
colitis. Gastroenterology 2001;121:805–11.
69. Meier C, Hegazi RA, Aisenberg J, et al. Innate immune receptor genetic polymorphisms in pouchitis: is NOD2/CARD15 a susceptibility factor? Inflamm
Bowel Dis 2005;11:965–71.
70. Schmidt CM, Lazenby AJ, Hendrickson RJ, et al. Pre-operative terminal ileal and
colonic resection histopathology predicts risk of pouchitis in patients after ileoanal pull-through procedure. Ann Surg 1998;227:654–62.
71. Achkar JP, Al-Haddad M, Lashner B, et al. Differentiating risk factors for acute
and chronic pouchitis. Clin Gastroenterol Hepatol 2005;3:60–6.
Ileal Pouch Disorders
72. Okon A, Dubinsky M, Vasilauskas EA, et al. Elevated platelet count before ileal
pouch–anal anastomosis for ulcerative colitis is associated with the development of chronic pouchitis. Am Surg 2005;71:821–6.
73. Hata K, Watanabe T, Shinozaki M, et al. Patients with extraintestinal manifestations have a higher risk of developing pouchitis in ulcerative colitis – multivariate
analysis. Scand J Gastroenterol 2003;38:1055–8.
74. Lepisto¨ A, Ka¨rkka¨inen P, Ja¨rvinen HJ. Prevalence of primary sclerosing cholangitis in ulcerative colitis patients undergoing proctocolectomy and ileal pouchanal anastomosis. Inflamm Bowel Dis 2008;14:775–9.
75. Shen B, Fazio VW, Remzi FH, et al. Risk factors for diseases of ileal pouch-anal
anastomosis in patients with ulcerative colitis. Clin Gastroenterol Hepatol 2006;
4:81–9.
76. Fleshner PR, Vasiliauskas EA, Kam LY, et al. High level perinuclear antineutrophil
cytoplasmic antibody (pANCA) in ulcerative colitis patients before colectomy
predicts the development of chronic pouchitis after ileal pouch-anal anastomosis. Gut 2001;49:671–7.
77. Kuisma J, Jarvinen H, Kahri A, et al. Factors associated with disease activity of
pouchitis after surgery for ulcerative colitis. Scand J Gastroenterol 2004;39:
544–8.
78. Hoda KM, Collins JF, Knigge KL, et al. Predictors of pouchitis after ileal pouchanal anastomosis: a retrospective review. Dis Colon Rectum 2008;51:554–60.
79. Fleshner P, Ippoliti A, Dubinsky M, et al. A prospective multivariate analysis of
clinical factors associated with pouchitis after ileal pouch-anal anastomosis.
Clin Gastroenterol Hepatol 2007;5:952–8.
80. Boyko EJ, Koepsell TD, Perera DR, et al. Risk of ulcerative colitis among former
and current cigarette smokers. N Engl J Med 1987;316:707–10.
81. Shen B, Remzi FH, Bennett AE, et al. Association between immune-associated
disorders and adverse outcomes of ileal pouch-anal anastomosis. Am J Gastroenterol 2009;104:655–64.
82. Mathis KL, Dozois EJ, Larson DW, et al. Ileal pouch-anal anastomosis and liver
transplantation for ulcerative colitis complicated by primary sclerosing cholangitis. Br J Surg 2008;95:882–6.
83. Cho CS, Dayton MT, Thompson JS, et al. Proctocolectomy-ileal pouch-anal
anastomosis for ulcerative colitis after liver transplantation for primary sclerosing cholangitis: a multi-institutional analysis. J Gastrointest Surg 2008;12:
1221–6.
84. Zins BJ, Sandborn WJ, Penna CR, et al. Pouchitis disease course after orthotopic liver transplantation in patients with primary sclerosing cholangitis and
an ileal pouch-anal anastomosis. Am J Gastroenterol 1995;90:2177–80.
85. Freeman K, Shao Z, Remzi FH, et al. Orthotopic liver transplantation for primary
sclerosing cholangitis in patients with ulcerative colitis: impact on occurrence of
chronic pouchitis. Clin Gastroenterol Hepatol 2008;6:62–8.
86. Shen B, Achkar J-P, Lashner BA, et al. Endoscopic and histologic evaluations
together with symptom assessment are required to diagnose pouchitis. Gastroenterology 2001;121:261–7.
87. Moskowitz RL, Shepherd NA, Nicholls RJ. An assessment of inflammation in the
reservoir after restorative proctocolectomy with ileoanal ileal reservoir. Int J Colorectal Dis 1986;1:167–74.
88. Sandborn WJ, Tremaine WJ, Batts KP, et al. Pouchitis after ileal pouch-anal
anastomosis: a pouchitis disease activity index. Mayo Clin Proc 1994;69:
409–15.
665
666
Wu & Shen
89. Shen B, Fazio VW, Bennett AE, et al. Effect of withdrawal of non-steroidal antiinflammatory drug use in patients with the ileal pouch. Dig Dis Sci 2007;52:
3321–8.
90. Parsi MA, Shen B, Achkar JP, et al. Fecal lactoferrin for diagnosis of symptomatic
patients with ileal pouch-anal anastomosis. Gastroenterology 2004;126:1280–6.
91. Lim M, Gonsalves S, Thekkinkattil D, et al. The assessment of a rapid noninvasive immunochromatographic assay test for fecal lactoferrin in patients with suspected inflammation of the ileal pouch. Dis Colon Rectum 2008;51:96–9.
92. Parsi MA, Ellis JJ, Lashner BA. Cost-effectiveness of quantitative fecal lactoferrin
assay for diagnosis of symptomatic patients with ileal pouch-anal anastomosis.
J Clin Gastroenterol 2008;42:799–805.
93. Johnson MW, Maestranzi S, Duffy AM, et al. Faecal calprotectin: a noninvasive
diagnostic tool and marker of severity in pouchitis. Eur J Gastroenterol Hepatol
2008;20:174–9.
94. Johnson MW, Maestranzi S, Duffy AM, et al. Faecal M2-pyruvate kinase: a novel,
noninvasive marker of ileal pouch inflammation. Eur J Gastroenterol Hepatol
2009;21:544–60.
95. Schmidt C, Ha¨user W, Giese T, et al. Irritable pouch syndrome is associated with
depressiveness and can be differentiated from pouchitis by quantification of
mucosal levels of proinflammatory gene transcripts. Inflamm Bowel Dis 2007;
13:1502–8.
96. Panis Y, Poupard B, Nemeth J, et al. Ileal pouch-anal anastomosis for Crohn’s
disease. Lancet 1996;347:854–7.
97. Shen B, Achkar J-P, Lashner BA, et al. Irritable pouch syndrome: a new category
of diagnosis for symptomatic patients with ileal pouch-anal anastomosis. Am J
Gastroenterol 2002;97:972–7.
98. Shen B, Liu W, Remzi FH, et al. Enterochromaffin cell hyperplasia in irritable
pouch syndrome. Am J Gastroenterol 2008;103:2293–300.
99. Lohmuller JL, Pemberton HJ, Dozois RR, et al. Pouchitis and extraintestinal
manifestations of inflammatory bowel disease after ileal pouch-anal anastomosis. Ann Surg 1990;211:622–9.
100. Mowschenson PM, Critchlow JF, Peppercorn MA. Ileoanal pouch operation: longterm outcome with or without diverting ileostomy. Arch Surg 2000;135:463–5.
101. Hurst RD, Chung TP, Rubin M, et al. Implications of acute pouchitis on the longterm functional results after restorative proctocolectomy. Inflamm Bowel Dis
1998;4:280–4.
102. Madiba TE, Bartolo DC. Pouchitis following restorative proctocolectomy for
ulcerative colitis: incidence and therapeutic outcome. J R Coll Surg Edinb
2001;46:334–7.
103. Sandborn WJ. Pouchitis: risk factors, frequency, natural history, classification
and public health prospective. In: McLeod RS, Martin F, Sutherland LR, et al,
editors. Trends in inflammatory bowel disease 1996. Lancaster (UK): Kluwer
Academic Publishers; 1997. p. 51–63.
104. Shen B. Diagnosis and management of patients with pouchitis. Drugs 2003;65:
453–61.
105. Croagh C, Shepherd SJ, Berryman M, et al. Pilot study on the effect of reducing
dietary FODMAP intake on bowel function in patients without a colon. Inflamm
Bowel Dis 2007;13:1522–8.
106. Stahlberg D, Gullberg K, Liljeqvist L, et al. Pouchitis following pelvic pouch operation for ulcerative colitis. Incidence, cumulative risk, and risk factors. Dis Colon
Rectum 1996;39:1012–8.
Ileal Pouch Disorders
107. Elahi B, Nikfar S, Derakhshani S, et al. On the benefit of probiotics in the
management of pouchitis in patients underwent ileal pouch anal anastomosis:
a meta-analysis of controlled clinical trials. Dig Dis Sci 2008;53:1278–84.
108. Gosselink MP, Schouten WR, van Lieshout LMC, et al. Delay of the first onset of
pouchitis by oral intake of the probiotic strain Lactobacillus rhamnosus GG. Dis
Colon Rectum 2004;47:876–84.
109. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as maintenance
treatment in patients with chronic pouchitis: a double-blind, placebo-controlled
trial. Gastroenterology 2000;119:305–9.
110. Mimura T, Rizzello F, Helwig U, et al. Once daily high dose probiotic therapy
(VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut
2004;53:108–14.
111. Pronio A, Montesani C, Butteroni C, et al. Probiotic administration in patients with
ileal pouch-anal anastomosis for ulcerative colitis is associated with expansion
of mucosal regulatory cells. Inflamm Bowel Dis 2008;14:662–8.
112. Gionchetti P, Rizzello F, Morselli C, et al. High-dose probiotics for the treatment
of active pouchitis. Dis Colon Rectum 2007;50:2075–82 [discussion: 2082–4].
113. Shen B, Brzezinski A, Fazio VW, et al. Maintenance therapy with a probiotic in
antibiotic-dependent pouchitis – experience in clinical practice. Aliment
Pharmacol Ther 2005;22:721–8.
114. McLaughlin SD, Johnson MW, Clark SK, et al. VSL#3 for chronic pouchitis;
experience in UK clinical practice [abstract]. Gastroenterology 2008;
134(Suppl 1):A711.
115. Madden MV, McIntyre AS, Nicholls RJ. Double-blinded crossover trial of metronidazole versus placebo in chronic unremitting pouchitis. Dig Dis Sci 1994;39:1193–6.
116. Shen B, Achkar JP, Lashner BA, et al. A randomized trial of ciprofloxacin and
metronidazole in treating acute pouchitis. Inflamm Bowel Dis 2001;7:301–5.
117. Isaacs KL, Sandler RS, Abreu M, et al. Crohn’s and Colitis Foundation of America
Clinical Alliance. Rifaximin for the treatment of active pouchitis: a randomized,
double-blind, placebo-controlled pilot study. Inflamm Bowel Dis 2007;13:1250–5.
118. McLaughlin SD, Clark SK, Bell AJ, et al. An open study of antibiotics for the
treatment of pre-pouch ileitis following restorative proctocolectomy with ileal
pouch-anal anastomosis. Aliment Pharmacol Ther 2008 Oct 3 [Epub ahead print].
119. Araki Y, Mitsuyama K, Nagae T, et al. Leukocytapheresis for the treatment of
active pouchitis: a pilot study. J Gastroenterol 2008;43:571–5.
120. Shen B, Pardi DS, Bennett AE, et-al. The efficacy and tolerability of AST-120
(spherical carbon adsorbent) in active pouchitis. Am J Gastroenterol
2009;104:1468–74.
121. Shen B, Remzi FH, Lopez AR, et al. Rifaximin for maintenance therapy in antibiotic-dependent pouchitis [abstract]. BMC Gastroenterol 2008;8:26.
122. Schaus BJ, Fazio VW, Remzi FH, et al. Large polyps in the ileal pouch in patients
with underlying ulcerative colitis. Dis Colon Rectum 2007;50:832–8.
123. Gionchetti P, Rizzello F, Venturi A, et al. Antibiotic combination therapy in
patients with chronic treatment-resistant pouchitis. Aliment Pharmacol Ther
1999;13:713–8.
124. Abdelrazeq AS, Kelly SM, Lund JN, et al. Rifaximin-ciprofloxacin combination
therapy is effective in chronic active refractory pouchitis. Colorectal Dis 2005;
7:182–6.
125. Mimura T, Rizzello R, Helwig U, et al. Four-week open-label trial of metronidazole
and ciprofloxacin for the treatment of recurrent or refractory pouchitis. Aliment
Pharmacol Ther 2002;16:909–17.
667
668
Wu & Shen
126. Shen B, Fazio VW, Remzi FH, et al. Combined ciprofloxacin and tinidazole in the
treatment of chronic refractory pouchitis. Dis Colon Rectum 2007;50:498–508.
127. Viscido A, Kohn A, Papi C, et al. Management of refractory fistulizing pouchitis
with infliximab. Eur Rev Med Pharmacol Sci 2004;8:239–46.
128. Gionchetti P, Rizzello F, Poggioli G, et al. Oral budesonide in the treatment of
chronic refractory pouchitis. Aliment Pharmacol Ther 2007;25:1231–6.
129. Calabrese C, Gionchetti P, Rizzello F, et al. Short-term treatment with infliximab
in chronic refractory pouchitis and ileitis. Aliment Pharmacol Ther 2008;27:
759–64.
130. Tulchinsky H, Dotan I, Alper A, et al. Comprehensive pouch clinic concept for
follow-up of patients after ileal pouch anal anastomosis: report of 3 years’ experience in a tertiary referral center. Inflamm Bowel Dis 2008;14:1125–32.